Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 344
Filtrar
1.
J Neuroendocrinol ; 32(7): e12877, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32572994

RESUMO

Neurokinin B (NKB) is critical for fertility in humans and stimulates gonadotrophin-releasing hormone/luteinising hormone (LH) secretion in several species, including sheep. There is increasing evidence that the actions of NKB in the retrochiasmatic area (RCh) contribute to the induction of the preovulatory LH surge in sheep. In the present study, we determined whether there are sex differences in the response to RCh administration of senktide, an agonist to the NKB receptor (neurokinin receptor-3 [NK3R]), and in NKB and NK3R expression in the RCh of sheep. To normalise endogenous hormone concentrations, animals were gonadectomised and given implants to mimic the pattern of ovarian steroids seen in the oestrous cycle. In females, senktide microimplants in the RCh produced an increase in LH concentrations that lasted for at least 8 hours after the start of treatment, whereas a much shorter increment (approximately 2 hours) was seen in males. We next collected tissue from gonadectomised lambs 18 hours after the insertion of oestradiol implants that produce an LH surge in female, but not male, sheep for immunohistochemical analysis of NKB and NK3R expression. As expected, there were more NKB-containing neurones in the arcuate nucleus of females than males. Interestingly, there was a similar sexual dimorphism in NK3R-containing neurones in the RCh, NKB-containing close contacts onto these RCh NK3R neurones, and overall NKB-positive fibres in this region. These data demonstrate that there are both functional and morphological sex differences in NKB-NK3R signalling in the RCh and raise the possibility that this dimorphism contributes to the sex-dependent ability of oestradiol to induce an LH surge in female sheep.


Assuntos
Hipotálamo Médio/metabolismo , Neurocinina B/metabolismo , Caracteres Sexuais , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Kisspeptinas/metabolismo , Masculino , Neurônios/metabolismo , Receptores de Taquicininas/metabolismo , Ovinos , Transdução de Sinais/fisiologia
2.
FASEB J ; 34(5): 7058-7074, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32275331

RESUMO

The role of central juxtaposed with another zinc finger gene 1 (JAZF1) in glucose regulation remains unclear. Here, we activated mediobasal hypothalamus (MBH) JAZF1 in high-fat diet (HFD)-fed rats by an adenovirus expressing JAZF1 (Ad-JAZF1). We evaluated the changes in the hypothalamic insulin receptor (InsR)-PI3K-Akt-AMPK pathway and hepatic glucose production (HGP). To investigate the impact of MBH Ad-JAZF1 on HGP, we activated MBH JAZF1 in the presence or absence of ATP-dependent potassium (KATP ) channel inhibition, hepatic branch vagotomy (HVG), or an AMPK activator (AICAR). In HFD-fed rats, MBH Ad-JAZF1 decreased body weight and food intake, and inhibited HGP by increasing hepatic insulin signaling. Under insulin stimulation, MBH Ad-JAZF1 increased InsR and Akt phosphorylation, and phosphatidylinositol 3, 4, 5-trisphosphate (PIP3) formation; however, AMPK phosphorylation was decreased in the hypothalamus. The positive effect of MBH JAZF1 on hepatic insulin signaling and HGP was prevented by treatment with a KATP channel inhibitor or HVG. The metabolic impact of hypothalamic JAZF1 was also blocked by MBH AICAR. Ad-JAZF1 treatment in SH-SY5Y cells resulted in an elevation of InsR and Akt phosphorylation following insulin stimulation. Our findings show that hypothalamic JAZF1 regulates HGP via the InsR-PI3K-Akt-AMPK pathway and KATP channels.


Assuntos
Proteínas Correpressoras/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose/biossíntese , Hipotálamo Médio/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Proteínas Correpressoras/genética , Proteínas de Ligação a DNA/genética , Dieta Hiperlipídica , Gluconeogênese , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Resistência à Insulina , Fígado/inervação , Fígado/metabolismo , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Nervo Vago/metabolismo
3.
J Neuroendocrinol ; 31(12): e12810, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31715027

RESUMO

The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.


Assuntos
Dinorfinas/metabolismo , Hipotálamo Médio/metabolismo , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Médio/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/farmacologia , Microinjeções , Neurocinina B/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Ratos , Receptor IGF Tipo 1/antagonistas & inibidores , Receptores da Neurocinina-3/biossíntese , Receptores Opioides/biossíntese , Maturidade Sexual , Substância P/análogos & derivados , Substância P/farmacologia , Regulação para Cima
4.
Am J Reprod Immunol ; 82(2): e13146, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31206871

RESUMO

PROBLEM: Current methods to quantify kisspeptin (KP) are limited. To this end, a radioimmunoassay (RIA) specific for KP was developed and validated. We hypothesized that use of a RIA would reveal multiple hypothalamic regions as targets of negative seasonal feedback of estradiol on KP production in sheep. METHOD OF STUDY: Ovariectomized (OVX) ewes bearing a subcutaneous implant of estradiol were euthanized during the breeding season (BS) (n = 4) and non-breeding season (NBS) (n = 3). Coronal sections of preoptic area (POA), anterior hypothalamic area (AHA), and mediobasal hypothalamus (MBH) were collected, as well as the median eminence (ME), cortex, brain stem, and cerebellum. Amounts of KP and gonadotropin-releasing hormone (GnRH) in individual hypothalamic nuclei were quantified by radioimmunoassay. RESULTS: Concentration and content of KP were lower during the NBS than the BS in the MBH (P < 0.01) and POA (P < 0.01). Levels of KP in tissue adjacent to the POA and MBH were much lower, and neither concentration nor content of KP differed between the BS and NBS. Kisspeptin was also detected in the cortex, brain stem, and cerebellum, but concentrations were not affected by season. In addition, concentration and content of GnRH in the POA, AHA, MBH, and ME were similar between seasons. CONCLUSION: Our RIA results indicate that in addition to the MBH, the POA and AHA appear to be involved in the seasonal negative feedback of estradiol on KP expression.


Assuntos
Córtex Cerebelar/metabolismo , Estradiol/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Médio/metabolismo , Kisspeptinas/metabolismo , Área Pré-Óptica/metabolismo , Ovinos/metabolismo , Animais , Cruzamento , Retroalimentação Fisiológica/fisiologia , Feminino , Radioimunoensaio , Estações do Ano
5.
Am J Physiol Endocrinol Metab ; 316(5): E948-E955, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30888861

RESUMO

Increased GABAergic output in the ventromedial hypothalamus (VMH) contributes to counterregulatory failure in recurrently hypoglycemic (RH) rats, and lactate, an alternate fuel source in the brain, contributes to this phenomenon. The current study assessed whether recurring bouts of glucose deprivation enhanced neuronal lactate uptake and, if so, whether this influenced γ-aminobutyric acid (GABA) output and the counterregulatory responses. Glucose deprivation was induced using 5-thioglucose (5TG). Control rats received an infusion of artificial extracellular fluid. These groups were compared with RH animals. Subsequently, the rats underwent a hypoglycemic clamp with microdialysis. To test whether 5TG affected neuronal lactate utilization, a subgroup of 5TG-treated rats was microinjected with a lactate transporter inhibitor [cyano-4-hydroxycinnamate (4CIN)] just before the start of the clamp. Both RH and 5TG raised VMH GABA levels, and this was associated with impaired counterregulatory responses. 4CIN reduced VMH GABA levels and restored the hormone responses in the 5TG group. We then evaluated [14C]lactate uptake in hypothalamic neuronal cultures. Recurring exposure to low glucose increased monocarboxylate transporter-2 mRNA expression and augmented lactate uptake. Taken together, our data suggest that glucose deprivation, per se, enhances lactate utilization in hypothalamic neurons, and this may contribute to suppression of the counterregulatory responses to hypoglycemia.


Assuntos
Glucose/metabolismo , Hipoglicemia/metabolismo , Hipotálamo Médio/citologia , Ácido Láctico/metabolismo , Neurônios/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Radioisótopos de Carbono , Catecolaminas/metabolismo , Ácidos Cumáricos/farmacologia , Glucose/análogos & derivados , Glucose/deficiência , Glucose/farmacologia , Técnica Clamp de Glucose , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Microdiálise , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/efeitos dos fármacos , Transportadores de Ácidos Monocarboxílicos/genética , Neurônios/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ácido gama-Aminobutírico/efeitos dos fármacos
6.
J Neuroendocrinol ; 31(2): e12686, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30633838

RESUMO

Nitric oxide (NO) negatively modulates the secretion of vasopressin (AVP), oxytocin (OT) and atrial natriuretic peptide (ANP) induced by the increase in extracellular osmolality, whereas carbon monoxide (CO) and hydrogen sulphide (H2 S) act to potentiate it; however, little information is available for the osmotic challenge model about whether and how such gaseous systems modulate each other. Therefore, using an acute ex vivo model of hypothalamic and neurohypophyseal explants (obtained from male 6/7-week-old Wistar rats) under conditions of extracellular iso- and hypertonicity, we determined the effects of NO (600 µmol L-1 sodium nitroprusside), CO (100 µmol L-1 tricarbonylchloro[glycinato]ruthenium [II]) and H2 S (10 mmol L-1 sodium sulphide) donors and nitric oxide synthase (NOS) (300 µmol L-1 Nω -methyl-l-arginine [LNMMA]), haeme oxygenase (HO) (200 µmol L-1 Zn(II) deuteroporphyrin IX 2,4-bis-ethylene glycol [ZnDPBG]) and cystathionine ß-synthase (CBS) (100 µmol L-1 aminooxyacetate [AOA]) inhibitors on the release of hypothalamic ANP and hypothalamic and neurohypophyseal AVP and OT, as well as on the activities of NOS, HO and CBS. LNMMA reversed hyperosmolality-induced NOS activity, and enhanced hormonal release by the hypothalamus and neurohypophysis, in addition to increasing CBS and hypothalamic HO activity. AOA decreased hypothalamic and neurohypophyseal CBS activity and hormonal release, whereas ZnDPBG inhibited HO activity and hypothalamic hormone release; however, in both cases, AOA did not modulate NOS and HO activity and ZnDPBG did not affect NOS and CBS activity. Thus, our data indicate that, although endogenous CO and H2 S positively modulate AVP, OT and ANP release, only NO plays a concomitant role of modulator of hormonal release and CBS activity in the hypothalamus and neurohypophysis and that of HO activity in the hypothalamus during an acute osmotic stimulus, which suggests that NO is a key gaseous controller of the neuroendocrine system.


Assuntos
Fator Natriurético Atrial/metabolismo , Monóxido de Carbono/metabolismo , Sulfeto de Hidrogênio/metabolismo , Hipotálamo Médio/metabolismo , Óxido Nítrico/metabolismo , Ocitocina/metabolismo , Vasopressinas/metabolismo , Animais , Cistationina beta-Sintase/metabolismo , Masculino , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Ratos Wistar , Sulfurtransferases/metabolismo
7.
Endocrinology ; 159(1): 145-162, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29145563

RESUMO

Aberrant neuronal DNA methylation patterns have been implicated in the promotion of obesity development; however, the role of neuronal DNA methyltransferases (Dnmts), enzymes that catalyze DNA methylation, in energy balance remains poorly understood. We investigated whether neuronal Dnmt1 regulates normal energy homeostasis and obesity development using a neuronal Dnmt1 knockout (ND1KO) mouse model, Dnmt1fl/fl Synapsin1Cre, which specifically deletes Dnmt1 in neurons. Neuronal Dnmt1 deficiency reduced adiposity in chow-fed mice and attenuated obesity in high-fat diet (HFD)-fed male mice. ND1KO male mice had reduced food intake and increased energy expenditure with the HFD. Furthermore, these mice had improved insulin sensitivity, as measured using an insulin tolerance test. The HFD-fed ND1KO mice had smaller fat pads and upregulation of thermogenic genes in brown adipose tissue. These data suggest that neuronal Dnmt1 plays an important role in regulating energy homeostasis. Notably, ND1KO male mice had elevated estrogen receptor-α (ERα) gene expression in the medial hypothalamus, which previously has been shown to control body weight. Immunohistochemistry experiments revealed that ERα protein expression was upregulated specifically in the dorsomedial region of the ventromedial hypothalamus, a region that might mediate the central effect of leptin. We conclude that neuronal Dnmt1 regulates energy homeostasis through pathways controlling food intake and energy expenditure. In addition, ERα expression in the dorsomedial region of the ventromedial hypothalamus might mediate these effects.


Assuntos
Adiposidade , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metabolismo Energético , Hipotálamo Médio , Proteínas do Tecido Nervoso/metabolismo , Neurônios/enzimologia , Obesidade/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/patologia , Animais , Cruzamentos Genéticos , DNA (Citosina-5-)-Metiltransferase 1/deficiência , DNA (Citosina-5-)-Metiltransferase 1/genética , Metilação de DNA , Dieta Hiperlipídica/efeitos adversos , Ingestão de Energia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica , Hipotálamo Médio/enzimologia , Hipotálamo Médio/metabolismo , Hipotálamo Médio/patologia , Resistência à Insulina , Masculino , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neurônios/patologia , Obesidade/etiologia , Obesidade/patologia , Obesidade/prevenção & controle , Regiões Promotoras Genéticas , Caracteres Sexuais
8.
Nat Commun ; 8: 15143, 2017 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-28489068

RESUMO

Consuming a calorically dense diet stimulates microglial reactivity in the mediobasal hypothalamus (MBH) in association with decreased number of appetite-curbing pro-opiomelanocortin (POMC) neurons; whether the reduction in POMC neuronal function is secondary to the microglial activation is unclear. Here we show that in hypercaloric diet-induced obese mice, persistently activated microglia in the MBH hypersecrete TNFα that in turn stimulate mitochondrial ATP production in POMC neurons, promoting mitochondrial fusion in their neurites, and increasing POMC neuronal firing rates and excitability. Specific disruption of the gene expressions of TNFα downstream signals TNFSF11A or NDUFAB1 in the MBH of diet-induced obese mice reverses mitochondrial elongation and reduces obesity. These data imply that in a hypercaloric environment, persistent elevation of microglial reactivity and consequent TNFα secretion induces mitochondrial stress in POMC neurons that contributes to the development of obesity.


Assuntos
Hipotálamo Médio/metabolismo , Microglia/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina , Estresse Fisiológico , Fator de Necrose Tumoral alfa/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Complexo I de Transporte de Elétrons/genética , Regulação da Expressão Gênica , Camundongos , Dinâmica Mitocondrial , Neuritos/metabolismo , Ligante RANK/genética , Transdução de Sinais
9.
Mol Metab ; 6(3): 306-312, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28271037

RESUMO

OBJECTIVE: Central cholinergic neural circuits play a role in the regulation of feeding behavior. The dorsomedial hypothalamus (DMH) is considered the appetite-stimulating center and contains cholinergic neurons. Here, we study the role of DMH cholinergic neurons in the control of food intake. METHODS: To selectively stimulate DMH cholinergic neurons, we expressed stimulatory designer receptors exclusively activated by designer drugs (DREADDs) and channelrhodopsins in DMH cholinergic neurons by injection of adeno-associated virus (AAV) vectors into the DMH of choline acetyltransferase (ChAT)-IRES-Cre mice. We also generated transgenic mice expressing channelrhodopsins in cholinergic neurons with the Cre-LoxP technique. To delete the Chat gene exclusively in the DMH, we injected an AAV carrying a Cre recombinase transgene into the DMH of floxed ChAT mice. Food intake was measured with and without selective stimulation of DMH cholinergic neurons. RESULTS: Mice lacking the Chat gene in the DMH show reduced body weight as compared to control. Chemogenetic activation of DMH cholinergic neurons promotes food intake. This orexigenic effect is further supported by experiments of optogenetic stimulation of DMH cholinergic neurons. DMH cholinergic neurons innervate pro-opiomelanocortin neurons in the arcuate nucleus of the hypothalamus (ARC). Treatment with acetylcholine (ACh) enhances GABAergic inhibitory transmission to ARC POMC neurons that is blocked by the muscarinic receptor antagonist. Direct activation of cholinergic fibers in the ARC readily stimulates food intake that is also abolished by the muscarinic receptor antagonist. CONCLUSION: ACh released from DMH cholinergic neurons regulates food intake and body weight. This effect is mediated in part through regulation of ARC POMC neurons. Activation of muscarinic receptors on GABAergic axon terminals enhances inhibitory tone to ARC POMC neurons. Hence, this novel DMHACh â†’ ARCPOMC pathway plays an important role in the control of food intake and body weight.


Assuntos
Regulação do Apetite/fisiologia , Neurônios Colinérgicos/fisiologia , Hipotálamo Médio/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/metabolismo , Dependovirus/genética , Núcleo Hipotalâmico Dorsomedial/metabolismo , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Hipotálamo Médio/metabolismo , Integrases , Leptina/genética , Masculino , Camundongos , Camundongos Transgênicos , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
10.
Physiol Behav ; 178: 93-102, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-27746261

RESUMO

Identifying the neurobiological mechanisms that underlie differential sensitivity to stress is critical for understanding the development and expression of stress-induced disorders, such as post-traumatic stress disorder (PTSD). Preclinical studies have suggested that rodents display different phenotypes associated with extinction of Pavlovian conditioned fear responses, with some rodent populations being resistant to extinction. An emerging literature also suggests a role for orexins in the consolidation processes associated with fear learning and extinction. To examine the possibility that the orexin system might be involved in individual differences in fear extinction, we used a Pavlovian conditioning paradigm in outbred Long-Evans rats. Rats showed significant variability in the extinction of cue-conditioned freezing and extinction recall, and animals were divided into groups based on their extinction profiles based on a median split of percent freezing behavior during repeated exposure to the conditioned cue. Animals resistant to extinction (high freezers) showed more freezing during repeated cue presentations during the within trial and between trial extinction sessions compared with the group showing significant extinction (low freezers), although there were no differences between these groups in freezing upon return to the conditioned context or during the conditioning session. Following the extinction recall session, activation of orexin neurons was determined using dual label immunohistochemistry for cFos in orexin positive neurons in the hypothalamus. Individual differences in the extinction of cue conditioned fear were associated with differential activation of hypothalamic orexin neurons. Animals showing poor extinction of cue-induced freezing (high freezers) had significantly greater percentage of orexin neurons with Fos in the medial hypothalamus than animals displaying significant extinction and good extinction recall (low freezers). Further, the freezing during extinction learning was positively correlated with the percentage of activated orexin neurons in both the lateral and medial hypothalamic regions. No differences in the overall density of orexin neurons or Fos activation were seen between extinction phenotypes. Although correlative, our results support other studies implicating a role of the orexinergic system in regulating extinction of conditioned responses to threat.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Hipotálamo Médio/metabolismo , Neurônios/metabolismo , Orexinas/metabolismo , Animais , Animais não Endogâmicos , Condicionamento Clássico/fisiologia , Sinais (Psicologia) , Eletrochoque , Reação de Congelamento Cataléptica/fisiologia , Hipotálamo Médio/citologia , Imuno-Histoquímica , Individualidade , Masculino , Rememoração Mental/fisiologia , Neurônios/citologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos Long-Evans
11.
Neurochem Res ; 41(12): 3344-3355, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27696120

RESUMO

Estrogen action is necessary for evidencing the stimulatory action of mifepristone and naloxone on prolactin (PRL) secretion during late pregnancy. Our aim is to determine the mechanism mediating this facilitator action of estrogens. To investigate the hypothalamic mechanisms involved in estrogen actions in PRL secretion at the end of pregnancy, we measured the effect of pretreatment with the estrogen antagonist tamoxifen on the expression of tyrosine hydroxylase (TH), hormone receptors (ERα and ß, PRs, PRLR(long)), and µ- and κ- opioid receptors (ORs) at mRNA (by semiquantitative RT-PCR) and protein (by western blot for TH, PRLR(long), ERα, PRs, µ- and ORs) levels in extracts of medial basal hypothalamus (MBH) and serum PRL, E2 and P4 levels (by RIA) in mifepristone- and naloxone-treated rats. Tamoxifen administration partially prevented PRL release induced by the combined treatment. TH expression diminished and ERα expression increased in mifepristone-treated rats at mRNA and protein levels and tamoxifen partially prevented these changes with no effect on PRs expression. Mifepristone increased PRLR(long) mRNA levels; this increase was blocked by tamoxifen. Combined tamoxifen and mifepristone treatment decreased µ- and k-ORs mRNA but not protein levels. In conclusion, E2 induces neuroadaptive mechanisms necessary to facilitate PRL release preceding delivery. Acting through ERα, E2 modulates hypothalamic dopaminergic neurons activity, regulating TH, µ- and κ-ORs and PRLR(long) expression, and is necessary for evidencing the effects of P4 withdrawal. Its presence on days 14 and 15 of pregnancy is crucial to facilitate the opioid system modulation of PRL secretion at the end of pregnancy in the rat.


Assuntos
Estradiol/metabolismo , Prenhez/fisiologia , Prolactina/metabolismo , Animais , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Hipotálamo Médio/metabolismo , Mifepristona/farmacologia , Naloxona/farmacologia , Gravidez , Prenhez/efeitos dos fármacos , Progesterona/metabolismo , Ratos Wistar , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Receptores de Progesterona/metabolismo , Receptores da Prolactina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Am J Physiol Regul Integr Comp Physiol ; 310(6): R476-80, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26739651

RESUMO

Adropin, a recently described peptide hormone produced in the brain and liver, has been reported to have physiologically relevant actions on glucose homeostasis and lipogenesis, and to exert significant effect on endothelial function. We describe a central nervous system action of adropin to inhibit water drinking and identify a potential adropin receptor, the orphan G protein-coupled receptor, GPR19. Reduction in GPR19 mRNA levels in medial basal hypothalamus of male rats resulted in the loss of the inhibitory effect of adropin on water deprivation-induced thirst. The identification of a novel brain action of adropin and a candidate receptor for the peptide should extend and accelerate the study of the potential therapeutic value of adropin or its mimetics for the treatment of metabolic disorders.


Assuntos
Proteínas Sanguíneas/farmacologia , Encéfalo/efeitos dos fármacos , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Peptídeos/farmacologia , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores de Neurotransmissores/efeitos dos fármacos , Animais , Pressão Arterial/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Injeções Intraventriculares , Masculino , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotransmissores/metabolismo , Sede/efeitos dos fármacos , Privação de Água
13.
Am J Physiol Regul Integr Comp Physiol ; 310(2): R134-42, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26561644

RESUMO

Neuropeptide Y (NPY) in the dorsomedial hypothalamus (DMH) plays an important role in the regulation of energy balance. While DMH NPY overexpression causes hyperphagia and obesity in rats, knockdown of NPY in the DMH via adeno-associated virus (AAV)-mediated RNAi (AAVshNPY) ameliorates these alterations. Whether this knockdown has a therapeutic effect on obesity and glycemic disorder has yet to be determined. The present study sought to test this potential using a rat model of high-fat diet (HFD)-induced obesity and insulin resistance, mimicking human obesity with impaired glucose homeostasis. Rats had ad libitum access to rodent regular chow (RC) or HFD. Six weeks later, an oral glucose tolerance test (OGTT) was performed for verifying HFD-induced glucose intolerance. After verification, obese rats received bilateral DMH injections of AAVshNPY or the control vector AAVshCTL, and OGTT and insulin tolerance test (ITT) were performed at 16 and 18 wk after viral injection (23 and 25 wk on HFD), respectively. Rats were killed at 26 wk on HFD. We found that AAVshCTL rats on HFD remained hyperphagic, obese, glucose intolerant, and insulin resistant relative to lean control RC-fed rats receiving DMH injection of AAVshCTL, whereas these alterations were reversed in NPY knockdown rats fed a HFD. NPY knockdown rats exhibited normal food intake, body weight, glucose tolerance, and insulin sensitivity, as seen in lean control rats. Together, these results demonstrate a therapeutic action of DMH NPY knockdown against obesity and impaired glucose homeostasis in rats, providing a potential target for the treatment of obesity and diabetes.


Assuntos
Dieta Hiperlipídica , Técnicas de Silenciamento de Genes , Intolerância à Glucose/terapia , Hipotálamo Médio/metabolismo , Neuropeptídeo Y/deficiência , Obesidade/terapia , Terapêutica com RNAi/métodos , Adiposidade , Animais , Glicemia/metabolismo , Peso Corporal , Dependovirus/genética , Modelos Animais de Doenças , Regulação para Baixo , Ingestão de Alimentos , Metabolismo Energético , Vetores Genéticos , Intolerância à Glucose/sangue , Intolerância à Glucose/genética , Intolerância à Glucose/fisiopatologia , Hipotálamo Médio/fisiopatologia , Insulina/sangue , Resistência à Insulina , Masculino , Neuropeptídeo Y/genética , Obesidade/sangue , Obesidade/genética , Obesidade/fisiopatologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Fatores de Tempo
14.
Neuroendocrinology ; 103(6): 711-23, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26580201

RESUMO

Substance P (SP) was recently reported to be expressed in human kisspeptin/neurokinin B/dynorphin (KNDy) neurons and to enhance KNDy neuron excitability in the mouse hypothalamus. We therefore examined (1) interactions of SP and kisspeptin in the mediobasal hypothalamus of adult male rhesus monkeys using immunofluorescence, and (2) the ability of SP to induce LH release in GnRH-primed, agonadal juvenile male monkeys. SP cell bodies were observed only occasionally in the arcuate nucleus (Arc), but more frequently dorsal to the Arc in the region of the premammillary nucleus. Castration resulted in an increase in the number of SP cell bodies in the Arc but not in the other regions. SP fibers innervated the Arc, where they were found in close apposition with kisspeptin perikarya in the periphery of this nucleus. Beaded SP axons projected to the median eminence, where they terminated in the external layer and intermingled with beaded kisspeptin axons. Colocalization of the two peptides, however, was not observed. Although close apposition between SP fibers and kisspeptin neurons suggest a role for SP in modulating GnRH pulse generator activity, i.v. injections of SP failed to elicit release of GnRH (as reflected by LH) in the juvenile monkey. Although the finding of structural interactions between SP and kisspeptin neurons is consistent with the notion that this tachykinin may be involved in regulating pulsatile GnRH release, the apparent absence of expression of SP in KNDy neurons suggests that this peptide is unlikely to be a fundamental component of the primate GnRH pulse generator.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Médio , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Peptídeos/administração & dosagem , Substância P/metabolismo , Administração Intravenosa , Animais , Castração , Relação Dose-Resposta a Droga , Hipotálamo Médio/citologia , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Macaca mulatta , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
15.
J Neuroendocrinol ; 28(2): 12336, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26568535

RESUMO

Prolonged stress and repeated activation of the hypothalamic-pituitary-adrenal axis can result in many sex-dependent behavioural and metabolic changes in rats, including alterations in feeding behaviour and reduced body weight. In adults, these effects of stress can be mimicked by corticosterone, a major output of the hypothalamic-pituitary-adrenal axis, and recapitulate the stress-induced sex difference, such that corticosterone-treated males show greater weight loss than females. Similar to adults, chronic stress during adolescence leads to reduced weight gain, particularly in males. However, it is currently unknown whether corticosterone mediates this somatic change and whether additional measures of neuroendocrine function are affected by chronic corticosterone exposure during adolescence in a sex-dependent manner. Therefore, we examined the effects of non-invasively administered corticosterone (150 or 300 µg/ml) in the drinking water of male and female rats throughout adolescent development (30-58 days of age). We found that adolescent animals exposed to chronic corticosterone gain significantly less weight than controls, which may be partly mediated by the effects of corticosterone on food consumption, fluid intake and gonadal hormone function. Our data further show that, despite similar circulating corticosterone levels, males demonstrate a greater sensitivity to these changes than females. We also found that Npy1 and Npy5 receptor mRNA expression, genes implicated in appetite regulation, was significantly reduced in the ventral medial hypothalamus of corticosterone-treated males and females compared to controls. Finally, parameters of gonadal function, such as plasma sex steroid concentrations and weight of reproductive tissues, were reduced by adolescent corticosterone treatment, although only in males. The data obtained in the present study indicate that chronic corticosterone exposure throughout adolescent development results in significant and sex-dependent somatic and neuroendocrine changes, and the results also provide an experimental framework for further investigating the impact of corticosterone on metabolic and neuroendocrine function during adolescence.


Assuntos
Peso Corporal/efeitos dos fármacos , Corticosterona/administração & dosagem , Corticosterona/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Neuropeptídeo Y/biossíntese , Receptores de Neuropeptídeos/biossíntese , Animais , Corticosterona/sangue , Relação Dose-Resposta a Droga , Estradiol/sangue , Feminino , Hormônios Esteroides Gonadais/sangue , Hipotálamo Médio/metabolismo , Masculino , Ratos , Glândulas Seminais/efeitos dos fármacos , Caracteres Sexuais , Testículo/efeitos dos fármacos , Testosterona/sangue , Útero/efeitos dos fármacos
16.
Int J Dev Neurosci ; 46: 115-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26315400

RESUMO

Thyrotropin-releasing hormone (TRH) synthesized in hypothalamic paraventricular nucleus directs hypothalamus-pituitary-thyroid (HPT) axis function, regulating thyrotropin release from adenohypophysis and thyroid hormones serum concentration. Pyroglutamyl aminopeptidase II (PPII), a Zn-dependent metallopeptidase located in adenohypophysis and medial-basal-hypothalamus degrades TRH released from the median eminence and participates in HPT axis function by regulating TRH-induced thyrotropin release from adenohypophysis. It is unknown whether dietary Zn deficiency down-regulates PPII. Our aim was to compare adenohypohyseal and medial-basal-hypothalamic PPII activity and expression of adult rats fed a Zn-deficient diet (2ppm) throughout their lifespan (DD), prenatally (DC) or after weaning (CD) vs. that of animals fed a control diet (20ppm:CC). Female rats consumed a Zn-deficient or control diet from two weeks before gestation and up to the end of lactation. We analyzed adenohypophyseal and medial-basal-hypothalamic PPII activity of dams and male offspring when adults; its relation to median eminence TRH, serum thyrotropin, leptin and thyroid hormones concentration. Offspring ate the same diet as their dams (CC, DD) or were switched from dietary regime after weaning (CD, DC) and until 2.5 months of age. DD males showed decreased adenohypophyseal and medial-basal-hypothalamic PPII activity, along with high thyrotropin serum concentration. Post-weaning Zn-deficiency (CD) decreased PPII activity only in adenohypophysis and increased thyrotropin circulating levels. Zn-replenishment (DC) normalized PPII activity in both regions and serum thyrotropin concentration. Adenohypophyseal PPII activity decreased and prolactin levels increased in Zn-deficient dams. We concluded that long-term changes in dietary Zn down-regulate PPII activity independently of T3, increasing thyrotropin serum concentration, overall resembling sub-clinical hypothyroidism.


Assuntos
Regulação para Baixo/fisiologia , Hipotálamo Médio/metabolismo , Zinco/deficiência , Fatores Etários , Aminopeptidases/deficiência , Aminopeptidases/genética , Análise de Variância , Animais , Regulação para Baixo/efeitos dos fármacos , Feminino , Lactação/efeitos dos fármacos , Masculino , Gravidez , Prolactina/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , RNA Mensageiro/metabolismo , Radioimunoensaio , Ratos , Ratos Wistar , Fatores Sexuais , Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/genética , Hormônio Liberador de Tireotropina/metabolismo , Zinco/administração & dosagem , Zinco/sangue
17.
Food Chem Toxicol ; 83: 10-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26032630

RESUMO

Perfluorooctane sulfonate (PFOS) is a fluorinated organic compound. This chemical is neurotoxic and can alter the pituitary secretion. This is an initial study aimed at knowing the toxic effects of high doses of PFOS on prolactin secretion and the possible mechanisms involved in these alterations. For that, adult male rats were orally treated with 3.0 and 6.0 mg of PFOS/kg body weight (b.w.)/day for 28 days. At the end of the treatment, the serum levels of prolactin and estradiol as well as the concentration of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA) and gamma-aminobutyric acid (GABA) were quantified in the anterior and in the mediobasal hypothalamus. PFOS, at the administered doses, reduced prolactin and estradiol secretion, increased the concentration of dopamine and GABA in the anterior hypothalamus, and decreased the ratios DOPAC/dopamine and HVA/dopamine in this same hypothalamic area. The outcomes reported in this study suggest that (1) high doses of PFOS inhibit prolactin secretion in adult male rats; (2) only the periventricular-hypophysial dopaminergic (PHDA) neurons seem to be involved in this inhibitory effect but not the tuberoinfundibular dopaminergic (TIDA) and the tuberohypophysial dopaminergic (THDA) systems; (3) GABAergic cells from the paraventricular and supraoptic nuclei could be partially responsible for the PFOS action on prolactin secretion; and finally (4) estradiol might take part in the inhibition exerted by elevated concentration of PFOS on prolactin release.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Disruptores Endócrinos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Hipotálamo Anterior/efeitos dos fármacos , Hipotálamo Médio/efeitos dos fármacos , Adeno-Hipófise/efeitos dos fármacos , Prolactina/antagonistas & inibidores , Ácidos Alcanossulfônicos/administração & dosagem , Animais , Dopamina/química , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Relação Dose-Resposta a Droga , Disruptores Endócrinos/administração & dosagem , Poluentes Ambientais/administração & dosagem , Estradiol/sangue , Estradiol/metabolismo , Fluorocarbonos/administração & dosagem , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Hipotálamo Anterior/metabolismo , Hipotálamo Médio/metabolismo , Masculino , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/metabolismo , Adeno-Hipófise/metabolismo , Prolactina/sangue , Prolactina/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Núcleo Supraóptico/efeitos dos fármacos , Núcleo Supraóptico/metabolismo , Testes de Toxicidade Subaguda , Ácido gama-Aminobutírico/química , Ácido gama-Aminobutírico/metabolismo
18.
Alcohol Clin Exp Res ; 38(10): 2572-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25335926

RESUMO

BACKGROUND: Insulin-like growth factor-1 (IGF-1) and transforming growth factor ß1 (TGFß1) are produced in hypothalamic astrocytes and facilitate luteinizing hormone-releasing hormone (LHRH) secretion. IGF-1 stimulates release by acting directly on the LHRH nerve terminals and both peptides act indirectly through specific plastic changes on glial/tanycyte processes that further support LHRH secretion. Because the relationship between these growth factors in the hypothalamus is not known, we assessed the ability of IGF-1 to induce TGFß1 synthesis and release and the actions of alcohol (ALC) on this mechanism prior to the onset of puberty. METHODS: Hypothalamic astrocytes were exposed to medium only, medium plus IGF-1 (200 ng/ml), or medium plus IGF-1 with 50 mM ALC. After 18 hours, media were collected and assayed for TGFß1. For the in vivo experiment, prepubertal female rats were administered either ALC (3 g/kg) or water via gastric gavage at 07:30 hours and at 11:30 hours. At 09:00 hours, saline or IGF-1 was administered into the third ventricle. Rats were killed at 15:00 hours and the medial basal hypothalamus (MBH) was collected for assessment of TGFß1, IGF-1 receptor (IGF-1R), and Akt. RESULTS: IGF-1 induced TGFß1 release (p < 0.01) from hypothalamic astrocytes in culture, an action blocked by ALC. In vivo, IGF-1 administration caused an increase in TGFß1 protein compared with controls (p < 0.05), an action blocked by ALC as well as a phosphatidylinositol 3 kinase/Akt inhibitor. IGF-1 stimulation also increased both total (p< 0.01) and phosphorylated (p)-IGF-1R (p < 0.05) protein levels, and phosphorylated (p)-Akt levels (p < 0.01), which were also blocked by ALC. CONCLUSIONS: This study shows that ALC blocks IGF-1 actions to stimulate synthesis and release of hypothalamic TGFß1, total and p-IGF-1R, and p-Akt levels further demonstrating the inhibitory actions of ALC on puberty-related events associated with hypothalamic LHRH release.


Assuntos
Etanol/farmacologia , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Maturidade Sexual , Fator de Crescimento Transformador beta1/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo Médio/citologia , Técnicas In Vitro , Modelos Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor IGF Tipo 1/metabolismo
19.
Alcohol Clin Exp Res ; 38(5): 1321-9, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24588206

RESUMO

BACKGROUND: Alcohol (ALC) diminishes gonadotropin-releasing hormone (GnRH) secretion and delays puberty. Glial transforming growth factor ß1 (TGFß1) plays a role in glial-neuronal communications facilitating prepubertal GnRH secretion. We assessed the effects of acute ALC administration on TGFß1-induced GnRH gene expression in the brain preoptic area (POA) and release of the peptide from the medial basal hypothalamus (MBH). Furthermore, we assessed actions and interactions of TGFß1 and ALC on an adhesion/signaling gene family involved in glial-neuronal communications. METHODS: Prepubertal female rats were administered ALC or water via gastric gavage at 7:30 am. At 9:00 am, saline or TGFß1 (100 ng/3 µl) was administered into the third ventricle. At 3:00 pm, the POA was removed and frozen for gene expression analysis and repeated for protein assessments. In another experiment, the MBH was removed from ALC-free rats. After equilibration, tissues were incubated in Locke's medium only or medium containing TGFß1 with or without 50 mM ALC for measurement of GnRH peptide released in vitro. RESULTS: TGFß1 induced GnRH gene expression in the POA, and this effect was blocked by ALC. We also described the presence and responsiveness of the TGFß1 receptor in the POA and showed that acute ALC exposure not only altered the TGFß1-induced increase in TGFß-R1 protein expression but also the activation of receptor-associated proteins, Smad2 and Smad3, key downstream components of the TGFß1 signaling pathway. Assessment of an adhesion/signaling family consisting of glial receptor protein tyrosine phosphatase beta and neuronal contactin-associated protein-1 (Caspr1) and contactin showed that the neuronal components were induced by TGFß1 and that ALC blocked these effects. Finally, TGFß1 was shown to induce release of the GnRH peptide in vitro, an action that was blocked by ALC. CONCLUSIONS: We have demonstrated glial-derived TGFß1 induces GnRH gene expression in the POA and stimulates release of the peptide from the MBH, actions necessary for driving the pubertal process. Importantly, ALC acted at both brain regions to block stimulatory effects of TGFß1. Furthermore, ALC altered neuronal components of an adhesion/signaling family previously shown to be expressed on GnRH neurons and implicated in glial-GnRH neuronal communications. These results further demonstrate detrimental effects of ALC at puberty.


Assuntos
Etanol/farmacologia , Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo Médio/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Animais , Interações Medicamentosas , Feminino , Expressão Gênica/efeitos dos fármacos , Hipotálamo Médio/metabolismo , Área Pré-Óptica/metabolismo , Puberdade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Endocrinology ; 155(5): 1874-86, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24484170

RESUMO

Kisspeptin regulates reproductive events, including puberty and ovulation, primarily via GnRH neurons. Prolonged treatment of prepubertal striped bass females with kisspeptin (Kiss) 1 or Kiss2 peptides failed to enhance puberty but suggested a gnrh-independent pituitary control pathway. Kiss2 inhibited, but Kiss1 stimulated, FShß expression and gonadal development, although hypophysiotropic gnrh1 and gnrh receptor expression remained unchanged. In situ hybridization and immunohistochemistry on brains and pituitaries revealed a differential plasticity between the 2 kisspeptin neurons. The differences were most pronounced at the prespawning phase in 2 regions along the path of gnrh1 axons: the nucleus lateralis tuberis (NLT) and the neurohypophysis. Kiss1 neurons appeared in the NLT and innervated the neurohypophysis of prespawning males and females, reaching Lh gonadotropes in the proximal pars distalis. Males, at all reproductive stages, had Kiss2 innervations in the NLT and the neurohypophysis, forming large axonal bundles in the former and intermingling with gnrh1 axons. Unlike in males, only preovulatory females had massive NLT-neurohypophysis staining of kiss2. Kiss2 neurons showed a distinct appearance in the NLT pars ventralis-equivalent region only in spawning zebrafish, indicating that this phenomenon is widespread. These results underscore the NLT as important nuclei for kisspeptin action in 2 facets: 1) kisspeptin-gnrh interaction, both kisspeptins are involved in the regulation of gnrh release, in a stage- and sex-dependent manner, especially at the prespawning phase; and 2) gnrh-independent effect of Kiss peptides on the pituitary, which together with the plastic nature of their neuronal projections to the pituitary implies that a direct gonadotropic regulation is plausible.


Assuntos
Bass/fisiologia , Proteínas de Peixes/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Kisspeptinas/metabolismo , Maturidade Sexual , Proteínas de Xenopus/metabolismo , Animais , Aquicultura , Axônios/efeitos dos fármacos , Axônios/metabolismo , Relação Dose-Resposta a Droga , Implantes de Medicamento , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Proteínas de Peixes/biossíntese , Proteínas de Peixes/genética , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/genética , Sistema Hipotálamo-Hipofisário/citologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Hipotálamo Médio/citologia , Hipotálamo Médio/efeitos dos fármacos , Hipotálamo Médio/crescimento & desenvolvimento , Hipotálamo Médio/metabolismo , Kisspeptinas/administração & dosagem , Kisspeptinas/farmacologia , Maryland , Neuro-Hipófise/citologia , Neuro-Hipófise/efeitos dos fármacos , Neuro-Hipófise/crescimento & desenvolvimento , Neuro-Hipófise/metabolismo , Maturidade Sexual/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas de Xenopus/administração & dosagem , Proteínas de Xenopus/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA